1
|
Rana A, Bhattacharya P, Ganguly S, Saha S, Naskar S, Ghosh S, Shaikh AR, Koley M, Saha S, Mukherjee SK. Individualized Homeopathic Medicinal Products in the Treatment of Post-COVID-19 Conditions: A Double-Blind, Randomized, Placebo-Controlled, Feasibility Trial. JOURNAL OF INTEGRATIVE AND COMPLEMENTARY MEDICINE 2025; 31:64-74. [PMID: 39321328 DOI: 10.1089/jicm.2024.0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Objectives: Evidence suggests that post-coronavirus disease 2019 (COVID-19) is associated with reduced health-related quality of life, and up to 80% of those infected with COVID-19 may experience these symptoms. The objective of the present study was to identify the effects of individualized homeopathic medicinal products (IHMPs) against placebos in postCOVID-19 conditions. Design: Double-blind, randomized (1:1), two parallel arms, placebo-controlled, feasibility trial. Setting: D. N. De Homoeopathic Medical College & Hospital, Kolkata, West Bengal, India. Subjects: Sixty participants with post-COVID-19 conditions. Interventions: Group verum (n = 30; IHMPs plus concomitant care) versus group control (n = 30; placebos plus concomitant care). Outcome Measures: Feasibility issues; primary-post-COVID-19 symptoms checklist; secondary-Measure Yourself Medical Outcomes Profile version 2 (MYMOP-2); all of them were measured at baseline, and monthly intervals, up to 3 months. The intention-to-treat sample was analyzed; group differences were reported using descriptive statistics: means, 95% confidence intervals (CIs), and between group effect sizes (Cohen's d). Results: Feasibility concerns showed promise; recruitment, retention, and attrition rates were 34.2%, 95%, and 5%, respectively. Group differences in both primary and secondary outcomes favored IHMPs against placebos: symptoms checklist score mean difference after 3 months: -4.2, 95% CI -4.9 to -3.4, d = 2.854 and MYMOP-2 mean difference after 3 months: -2.2, 95% CI -2.8 to -1.7, d = 2.082, respectively. Natrum muriaticum (11.7%), Pulsatilla nigricans (10%), Rhus toxicodendron (8.3%), and Calcarea carbonica (8.3%) were the most frequently prescribed remedies. Conclusions: IHMPs produced better results than placebos in reducing symptoms checklist scores and MYMOP-2 scores in the treatment of post-COVID-19 conditions. Definitive trials are warranted to confirm the findings.
Collapse
Affiliation(s)
- Abhijit Rana
- Department of Organon of Medicine and Homoeopathic Philosophy, D. N. De Homoeopathic Medical College and Hospital, Govt. of West Bengal, Kolkata, India
| | - Pulakendu Bhattacharya
- Department of Organon of Medicine and Homoeopathic Philosophy, D. N. De Homoeopathic Medical College and Hospital, Govt. of West Bengal, Kolkata, India
| | - Subhasish Ganguly
- Department of Organon of Medicine and Homoeopathic Philosophy, D. N. De Homoeopathic Medical College and Hospital, Govt. of West Bengal, Kolkata, India
| | - Sangita Saha
- Department of Organon of Medicine and Homoeopathic Philosophy, D. N. De Homoeopathic Medical College and Hospital, Govt. of West Bengal, Kolkata, India
| | - Satyajit Naskar
- Department of Organon of Medicine and Homoeopathic Philosophy, D. N. De Homoeopathic Medical College and Hospital, Govt. of West Bengal, Kolkata, India
| | - Shubhamoy Ghosh
- Department of Pathology and Microbiology, D. N. De Homoeopathic Medical College & Hospital, Govt. of West Bengal, Kolkata, India
| | - Abdur Rahaman Shaikh
- Department of Practice of Medicine, D. N. De Homoeopathic Medical College and Hospital, Govt. of West Bengal, Kolkata, India
| | - Munmun Koley
- Department of Homeopathy, East Bishnupur State Homoeopathic Dispensary, Chandi Daulatabad Block Primary Health Centre, Department of Health & Family Welfare, Govt. of West, Kolkata, India
| | - Subhranil Saha
- Department of Repertory, D. N. De Homoeopathic Medical College and Hospital, Govt. of West Bengal, Kolkata, India
| | - Shyamal Kumar Mukherjee
- Department of Community Medicine, D. N. De Homoeopathic Medical College and Hospital, Govt. of West Bengal, Kolkata, India
| |
Collapse
|
2
|
Zhang C, Gerzanich V, Cruz-Cosme R, Zhang J, Tsymbalyuk O, Tosun C, Sallapalli BT, Liu D, Keledjian K, Papadimitriou JC, Drachenberg CB, Nasr M, Zhang Y, Tang Q, Simard JM, Zhao RY. SARS-CoV-2 ORF3a induces COVID-19-associated kidney injury through HMGB1-mediated cytokine production. mBio 2024; 15:e0230824. [PMID: 39345136 PMCID: PMC11559048 DOI: 10.1128/mbio.02308-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 10/01/2024] Open
Abstract
The primary challenge posed by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is COVID-19-related mortality, often exacerbated by additional medical complications, such as COVID-19-associated kidney injuries (CAKIs). Up to half of COVID-19 patients experience kidney complications, with those facing acute respiratory failure and kidney injury having the worst overall prognosis. Despite the significant impact of CAKI on COVID-19-related mortality and its enduring effects in long COVID, the underlying causes and molecular mechanisms of CAKI remain elusive. In this study, we identified a functional relationship between the expression of the SARS-CoV-2 ORF3a protein and inflammation-driven apoptotic death of renal tubular epithelial cells in patients with CAKI. We demonstrate in vitro that ORF3a independently induces renal cell-specific apoptotic cell death, as evidenced by the elevation of kidney injury molecule-1 (KIM-1) and the activation of NF-kB-mediated proinflammatory cytokine (TNFα and IL-6) production. By examining kidney tissues of SARS-CoV-2-infected K18-ACE2 transgenic mice, we observed a similar correlation between ORF3a-induced cytopathic changes and kidney injury. This correlation was further validated through reconstitution of the ORF3a effects via direct adenoviral injection into mouse kidneys. Through medicinal analysis, we identified a natural compound, glycyrrhizin (GL4419), which not only blocks viral replication in renal cells, but also mitigates ORF3a-induced renal cell death by inhibiting activation of a high mobility group box 1 (HMGB1) protein, leading to a reduction of KIM-1. Moreover, ORF3a interacts with HMGB1. Overproduction or downregulation of hmgb1 expression results in correlative changes in renal cellular KIM-1 response and respective cytokine production, implicating a crucial role of HMGB1 in ORF3a-inflicted kidney injuries. Our data suggest a direct functional link between ORF3a and kidney injury, highlighting ORF3a as a unique therapeutic target contributing to CAKI. IMPORTANCE The major challenge of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during the pandemic is COVID-19-related mortality, which has tragically claimed millions of lives. COVID-19-associated morbidity and mortality are often exacerbated by pre-existing medical conditions, such as chronic kidney diseases (CKDs), or the development of acute kidney injury (AKI) due to COVID-19, collectively known as COVID-19-associated kidney injuries (CAKIs). Patients who experience acute respiratory failure with CAKI have the poorest clinical outcomes, including increased mortality. Despite these alarming clinical findings, there is a critical gap in our understanding of the underlying causes of CAKI. Our study establishes a direct correlation between the expression of the SARS-CoV-2 viral ORF3a protein and kidney injury induced by ORF3a linking to CAKI. This functional relationship was initially observed in our clinical studies of COVID-19 patients with AKI and was further validated through animal and in vitro cellular studies, either by expressing ORF3a alone or in the context of viral infection. By elucidating this functional relationship and its underlying mechanistic pathways, our research deepens the understanding of COVID-19-associated kidney diseases and presents potential therapeutic avenues to address the healthcare challenges faced by individuals with underlying conditions.
Collapse
Affiliation(s)
- Chenyu Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ruth Cruz-Cosme
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cigdem Tosun
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Dongxiao Liu
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John C. Papadimitriou
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cinthia B. Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mohamed Nasr
- Division of AIDS, NIAID, NIH, Drug Development and Clinical Sciences Branch, Bethesda, Maryland, USA
| | - Yanjin Zhang
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - J. Marc Simard
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, Maryland, USA
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Merhavy ZI, Junor T, Gonzalez A, Filippis SMD, Oveisitork S, Rivera E, Ndukwu I, Bhatara K. Long COVID: A Comprehensive Overview of the Signs and Symptoms across Multiple Organ Systems. Korean J Fam Med 2024; 45:305-316. [PMID: 39600184 PMCID: PMC11605154 DOI: 10.4082/kjfm.24.0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 11/29/2024] Open
Abstract
Long coronavirus disease (COVID), also known as the post-acute sequelae of coronavirus disease 2019 (COVID-19) (PASC), is a significant concern since the end of the COVID-19 pandemic, as it still manifests in individuals with persistent symptoms and complications beyond the acute phase of infection. Defining this disease is challenging, as it manifests as a spectrum of symptoms varying in severity among individuals who have previously tested positive for COVID-19. Long COVID is more prevalent in hospitalized COVID-19 patients and presents in various ways, ranging from pulmonary to extrapulmonary symptoms. This literature review examines the current body of research on long COVID with a focus on its effects on the cardiovascular, hematological, respiratory, renal, and neurological systems with systematically analyzed, peer-reviewed articles retrieved from the PubMed database. There have been several proposed pathophysiological mechanisms by which severe acute respiratory syndrome coronavirus 2 affects the aforementioned organ systems; however, research on the definite mechanisms is lacking, especially when considering the management of long COVID in the perioperative setting. The impact of post-COVID sequelae necessitates individualized management strategies tailored to each symptomatic profile, particularly in patients with comorbidities. The COVID-19 pandemic affected millions of people and had a profound impact on those who developed PASC, lowering their quality of life and increasing potential surgical risks. However, there is still uncertainty regarding the specific risk factors for long COVID and who is most susceptible to it. Further research is required to fill these gaps and explore potential avenues for preventing PASC.
Collapse
Affiliation(s)
| | - Tiana Junor
- University of Medicine and Health Sciences, Basseterre, St. Kitts & Nevis
| | - Aranice Gonzalez
- University of Medicine and Health Sciences, Basseterre, St. Kitts & Nevis
| | | | | | - Eliu Rivera
- University of Medicine and Health Sciences, Basseterre, St. Kitts & Nevis
| | - Ifeanyi Ndukwu
- University of Medicine and Health Sciences, Basseterre, St. Kitts & Nevis
| | - Kanika Bhatara
- Department of Family Medicine, Wayne State University School of Medicine, Rochester, MI, USA
| |
Collapse
|
4
|
Kolesnyk M, Stepanova N, Ostapenko T, Snisar L, Rysev A, Shifris I. Смертність у хворих на ХХН VГД з постковідним синдромом: багатоцентрове, проспективне, когортне дослідження. UKRAINIAN JOURNAL OF NEPHROLOGY AND DIALYSIS 2024:68-78. [DOI: 10.31450/ukrjnd.3(83).2024.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Термінальна стадія хронічної хвороби нирок, яка потребує лікування методом гемодіалізу (ХХН VГД), є критичною глобальною проблемою охорони здоров'я, яка ще більше ускладнилась пандемією COVID-19. Хоча відомо, що постковідний синдром (ПКС) погіршує якість життя ГД пацієнтів, його вплив на смертність залишається маловивченим. Метою цього дослідження було проаналізувати смертність хворих на ХХН VГД з ПКС та визначити її клінічні та лабораторні детермінанти.
Методи. Це багатоцентрове, проспективне дослідження включало ГД пацієнтів з чотирьох діалізних центрів України, які перенесли COVID-19 станом на кінець березня 2022 року. Зібрані дані включали демографічні, клінічні та лабораторні параметри і наявність ПКС. Основними кінцевими точками були смертність у хворих, асоційована з ПКС або без нього.
Результати. З 465 ГД пацієнтів, 353 відповідали критеріям включення. ПКС був діагностований у 222 пацієнтів (62,9%), який асоціювався з чоловічою статтю, старшим віком, діабетом, тимчасовим судинним доступом, вищим індексом Чарлсона, нижчими показниками Kt/V, гемоглобіну та альбуміну, вищими балами CONUT та підвищеними рівнями СРБ. Протягом двох років спостереження зареєстровано 38 (11,4%) летальних випадків; основними причинами смерті були серцево-судинні події (42,2%) та інфекційні ускладнення (21,1%). Багатофакторний регресійний аналіз Кокса визначив, що старший вік (OR 1,32, 95% CI 1,03; 1,66), тривалість діалізу (OR 1,86, 95% CI 1,77; 1,97), діабет (OR 3,57, 95% CI 1,81; 7,06), вища коморбідність (OR 4,22, 95% CI 2,11; 7,82), гірший нутритивний статус (OR 1,66, 95% CI 1,08; 12,3), тимчасовий судинний доступ (OR 1,88, 95% CI 1,16; 3,86), високий артеріальний тиск (OR 1,37, 95% CI 1,03; 1,82) та рівень С-реактивного білка (OR 1,68, 95% CI 1,49; 1,95) і тяжкий перебіг COVID-19, який вимагав госпіталізації (OR 5,7, 95% CI 3,73; 7,89), є незалежними предикторами смертності. У той же час, цільові рівні гемоглобіну (OR 0,87, 95% CI 0,66; 0,99) та Kt/V (OR 0,74, 95% CI 0,68; 0,96), а також вакцинація проти COVID-19 (OR 0,17, 95% CI 0,05; 0,61) статистично значущо знижували імовірність смерті. Наявність ПКС не впливала на виживаність ГД пацієнтів протягом двох років.
Висновки. Дворічна смертність ГД пацієнтів з ПКС визначається комбінацією демографічних та клініко-лабораторних факторів, таких як старший вік, тривале лікування ГД, коморбідна патологія, білково-енергетична недостатність, тимчасовий судинний доступ, неадекватна діалізна доза, низькй рівень гемоглобіну, хронічне запалення, недосягнення цільового рівня артеріального тиску, негативний вакцинальний статус та тяжкий перебіг гострого періоду COVID-19 з необхідністю кисневої підтримки. Вакцинація та підтримання цільових рівнів показників моніторингу хворих на ХХН VГД після COVID-19 мають вирішальне значення для покращення виживання цієї категорії хворих.
Collapse
|
5
|
Coşkun A, Erdem K, Can U. Association between COVID-19 and increased incidence of overactive bladder symptoms in female patients: A retrospective analysis. Actas Urol Esp 2024; 48:454-460. [PMID: 38185318 DOI: 10.1016/j.acuroe.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 01/09/2024]
Abstract
INTRODUCTION AND OBJECTIVES To investigate female patients' post-COVID-19 voiding symptoms and to research how they relate to overactive bladder (OAB). PATIENTS One hundred and forty patients aged 20-50 years who were hospitalised and discharged due to COVID-19 at Kartal Dr. Lütfi Kırdar City Hospital between 2021 and 2022 and 50 patients with a history of COVID-19 among two hundred female patients who presented to the urology outpatient clinic with symptoms related to OAB were retrospectively analysed. Bladder diary, overactive bladder symptom score (OABSS), uroflowmetry values and time of onset of symptoms of symptomatic patients were recorded for all patients. Disease-free individuals for control purposes were not included in the study. RESULTS It was observed that 38% of 140 hospitalized patients had a symptomatic change related to OAB, and there was a significant difference in voiding diary, OABSS, and uroflowmetry Qmax values between symptomatic and non-symptomatic patients after COVID-19. (p:0.001) There was a significant difference between the pre-COVID-19 (estimated) and post-COVID-19 (current) voiding diary and OABSS values of all symptomatic patients (with and without a history of hospitalization). (p:0.001) When these two groups were compared with each other, there was a significant difference between the post-COVID-19 voiding diary, OABSS values, and the meantime to the onset of symptoms in inpatients and outpatients (p:0.001) CONCLUSION: COVID-19 may be associated with urgency/urge incontinence and overactive bladder in female patients.
Collapse
Affiliation(s)
- A Coşkun
- Servicio de Urología, Universidad de Ciencias de la Salud, Hospital Kartal Dr. Lutfi Kirdar, Estambul, Turkey.
| | - K Erdem
- Servicio de Urología, Hospital de Formación e Investigación en Medicina Física y Rehabilitación Ankara Gaziler, Ankara, Turkey
| | - U Can
- Servicio de Urología, Universidad de Ciencias de la Salud, Hospital Kartal Dr. Lutfi Kirdar, Estambul, Turkey
| |
Collapse
|
6
|
Boruga M, Septimiu-Radu S, Nandarge PS, Elagez A, Doros G, Lazureanu VE, Stoicescu ER, Tanase E, Iacob R, Dumitrescu A, Bota AV, Cotoraci C, Bratu ML. Kidney Function Tests and Continuous eGFR Decrease at Six Months after SARS-CoV-2 Infection in Patients Clinically Diagnosed with Post-COVID Syndrome. Biomedicines 2024; 12:950. [PMID: 38790912 PMCID: PMC11117851 DOI: 10.3390/biomedicines12050950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
The long-term sequelae of SARS-CoV-2 infection are still under research, since extensive studies showed plenty of systemic effects of the viral infection, extending even after the acute phase of the infection. This study evaluated kidney function tests six months after SARS-CoV-2 infection in patients clinically diagnosed with Post-COVID Syndrome, hypothesizing persistent renal dysfunction evidenced by altered kidney function tests compared to baseline levels. Continuous eGFR decrease <30 at six months post-infection was considered the main study outcome. Conducted at the "Victor Babes" Hospital, this retrospective observational study involved adults with laboratory-confirmed SARS-CoV-2 infection and clinically-diagnosed Post-COVID Syndrome, excluding those with prior chronic kidney disease or significant renal impairment. Kidney function tests, including serum creatinine, blood urea nitrogen (BUN), estimated glomerular filtration rate (eGFR), alongside markers of kidney damage such as proteinuria and hematuria, were analyzed. Among 206 participants, significant differences were observed between the control (n = 114) and the Post-COVID group (n = 92). The Post-COVID group exhibited higher serum creatinine (109.7 μmol/L vs. 84.5 μmol/L, p < 0.001), lower eGFR (65.3mL/min/1.73 m2 vs. 91.2 mL/min/1.73 m2, p < 0.001), and elevated BUN levels (23.7 mg/dL vs. 15.2 mg/dL, p < 0.001) compared to the control group. Regression analysis highlighted significant predictors of continuous eGFR decrease <30 at six months post-infection. The development of acute kidney injury (AKI) during the initial COVID-19 illness emerged as a strong predictor of reduced eGFR (β = 3.47, p < 0.001). Additional factors, including a creatinine increase (23 μmol/L above the normal range) and an elevated Albumin to Creatinine Ratio (ACR) (>11 mg/g above the normal range), were significantly associated with eGFR reduction. Patients with Post-COVID Syndrome demonstrate significant renal impairment six months post-SARS-CoV-2 infection. The study's findings stress the need for ongoing monitoring and intervention strategies for renal health in affected individuals, underscoring the persistent impact of COVID-19 on renal function.
Collapse
Affiliation(s)
- Madalina Boruga
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| | - Susa Septimiu-Radu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (E.R.S.); (E.T.); (R.I.); (M.L.B.)
| | - Prashant Sunil Nandarge
- Department of General Medicine, D.Y. Patil Medical College Kolhapur, Kolhapur 416005, India;
| | - Ahmed Elagez
- Department of General Medicine, Misr University for Science & Technology, Giza 3236101, Egypt;
| | - Gabriela Doros
- Third Discipline of Pediatrics, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| | - Voichita Elena Lazureanu
- Department XIII, Discipline of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| | - Emil Robert Stoicescu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (E.R.S.); (E.T.); (R.I.); (M.L.B.)
- Department of Radiology and Medical Imaging, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Elena Tanase
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (E.R.S.); (E.T.); (R.I.); (M.L.B.)
| | - Roxana Iacob
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (E.R.S.); (E.T.); (R.I.); (M.L.B.)
- Department of Anatomy and Embryology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | | | - Adrian Vasile Bota
- Multidisciplinary Doctoral School, “Vasile Goldis” Western University, 310025 Arad, Romania; (A.V.B.); (C.C.)
| | - Coralia Cotoraci
- Multidisciplinary Doctoral School, “Vasile Goldis” Western University, 310025 Arad, Romania; (A.V.B.); (C.C.)
- Department of Hematology, Faculty of Medicine, “Vasile Goldis” Western University, 310025 Arad, Romania
| | - Melania Lavinia Bratu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (E.R.S.); (E.T.); (R.I.); (M.L.B.)
- Center for Neuropsychology and Behavioral Medicine, Discipline of Psychology, Faculty of General Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Center for Cognitive Research in Neuropsychiatric Pathology, Department of Neurosciences, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| |
Collapse
|
7
|
Wang M, Li W, Wang H, Song P. Development and validation of machine learning-based models for predicting healthcare-associated bacterial/fungal infections among COVID-19 inpatients: a retrospective cohort study. Antimicrob Resist Infect Control 2024; 13:42. [PMID: 38616284 PMCID: PMC11017584 DOI: 10.1186/s13756-024-01392-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/30/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND COVID-19 and bacterial/fungal coinfections have posed significant challenges to human health. However, there is a lack of good tools for predicting coinfection risk to aid clinical work. OBJECTIVE We aimed to investigate the risk factors for bacterial/fungal coinfection among COVID-19 patients and to develop machine learning models to estimate the risk of coinfection. METHODS In this retrospective cohort study, we enrolled adult inpatients confirmed with COVID-19 in a tertiary hospital between January 1 and July 31, 2023, in China and collected baseline information at admission. All the data were randomly divided into a training set and a testing set at a ratio of 7:3. We developed the generalized linear and random forest models for coinfections in the training set and assessed the performance of the models in the testing set. Decision curve analysis was performed to evaluate the clinical applicability. RESULTS A total of 1244 patients were included in the training cohort with 62 healthcare-associated bacterial/fungal infections, while 534 were included in the testing cohort with 22 infections. We found that patients with comorbidities (diabetes, neurological disease) were at greater risk for coinfections than were those without comorbidities (OR = 2.78, 95%CI = 1.61-4.86; OR = 1.93, 95%CI = 1.11-3.35). An indwelling central venous catheter or urinary catheter was also associated with an increased risk (OR = 2.53, 95%CI = 1.39-4.64; OR = 2.28, 95%CI = 1.24-4.27) of coinfections. Patients with PCT > 0.5 ng/ml were 2.03 times (95%CI = 1.41-3.82) more likely to be infected. Interestingly, the risk of coinfection was also greater in patients with an IL-6 concentration < 10 pg/ml (OR = 1.69, 95%CI = 0.97-2.94). Patients with low baseline creatinine levels had a decreased risk of bacterial/fungal coinfections(OR = 0.40, 95%CI = 0.22-0.71). The generalized linear and random forest models demonstrated favorable receiver operating characteristic curves (ROC = 0.87, 95%CI = 0.80-0.94; ROC = 0.88, 95%CI = 0.82-0.93) with high accuracy, sensitivity and specificity of 0.86vs0.75, 0.82vs0.86, 0.87vs0.74, respectively. The corresponding calibration evaluation P statistics were 0.883 and 0.769. CONCLUSIONS Our machine learning models achieved strong predictive ability and may be effective clinical decision-support tools for identifying COVID-19 patients at risk for bacterial/fungal coinfection and guiding antibiotic administration. The levels of cytokines, such as IL-6, may affect the status of bacterial/fungal coinfection.
Collapse
Affiliation(s)
- Min Wang
- Department of Infection Management, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu Province, 210009, China
| | - Wenjuan Li
- Department of Medical Big Data, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu Province, 210009, China
| | - Hui Wang
- Department of Infection Management, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu Province, 210009, China
| | - Peixin Song
- Department of Infection Management, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School,Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu Province, 210009, China.
| |
Collapse
|
8
|
Arenas CL, Forero ACP, Ángel DCV, López PMR, Diaz LVG, Aguilar DKN, Yate HCM. Risk factors for acute kidney injury in patients hospitalized with COVID-19. J Bras Nefrol 2024; 46:e20230056. [PMID: 38078832 PMCID: PMC11210541 DOI: 10.1590/2175-8239-jbn-2023-0056en] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/20/2023] [Indexed: 03/21/2024] Open
Abstract
INTRODUCTION Acute kidney injury (AKI) occurs frequently in COVID-19 patients and is associated with greater morbidity and mortality. Knowing the risks of AKI allows for identification, prevention, and timely treatment. This study aimed to identify the risk factors associated with AKI in hospitalized patients. METHODS A descriptive, retrospective, cross-sectional, and analytical component study of adult patients hospitalized with COVID-19 from March 1 to December 31, 2020 was carried out. AKI was defined by the creatinine criteria of the KDIGO-AKI guidelines. Information, regarding risk factors, was obtained from electronic medical records. RESULTS Out of the 934 patients, 42.93% developed AKI, 60.59% KDIGO-1, and 9.9% required renal replacement therapy. Patients with AKI had longer hospital stay, higher mortality, and required more intensive care unit (ICU) admission, mechanical ventilation, and vasopressor support. Multivariate analysis showed that age (OR 1.03; 95% CI 1.02-1.04), male sex (OR 2.13; 95% CI 1.49-3.04), diabetes mellitus (DM) (OR 1.55; 95% CI 1.04-2.32), chronic kidney disease (CKD) (OR 2.07; 95% CI 1.06-4.04), C-reactive protein (CRP) (OR 1.02; 95% CI 1.00-1.03), ICU admission (OR 1.81; 95% CI 1.04-3.16), and vasopressor support (OR 7.46; 95% CI 3.34-16.64) were risk factors for AKI, and that bicarbonate (OR 0.89; 95% CI 0.84-0.94) and partial pressure arterial oxygen/inspired oxygen fraction index (OR 0.99; 95% CI 0.98-0.99) could be protective factors. CONCLUSIONS A high frequency of AKI was documented in COVID-19 patients, with several predictors: age, male sex, DM, CKD, CRP, ICU admission, and vasopressor support. AKI occurred more frequently in patients with higher disease severity and was associated with higher mortality and worse outcomes.
Collapse
|
9
|
Aklilu AM, Kumar S, Nugent J, Yamamoto Y, Coronel-Moreno C, Kadhim B, Faulkner SC, O’Connor KD, Yasmin F, Greenberg JH, Moledina DG, Testani JM, Wilson FP. COVID-19-Associated Acute Kidney Injury and Longitudinal Kidney Outcomes. JAMA Intern Med 2024; 184:414-423. [PMID: 38407862 PMCID: PMC10897825 DOI: 10.1001/jamainternmed.2023.8225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/08/2023] [Indexed: 02/27/2024]
Abstract
Importance COVID-19 infection is associated with a high incidence of acute kidney injury (AKI). Although rapid kidney function decline has been reported in the first few months after COVID-19-associated AKI (COVID-AKI), the longer-term association of COVID-AKI with kidney function remains unknown. Objective To assess long-term kidney outcomes of patients who had COVID-19-associated AKI. Design, Setting, and Participants This was a retrospective longitudinal multicenter cohort study conducted in a large hospital system using electronic health records data on adult hospitalized patients with AKI and COVID-19 or other illnesses. Included patients were hospitalized during the COVID-19 pandemic (March 2020-June 2022), were screened for SARS-CoV-2, had AKI, and survived to discharge, or had been hospitalized during the 5 years before the pandemic (October 2016-January 2020), had a positive influenza A or B test result, had AKI, and survived to discharge. Patients were followed up for a maximum of 2 years after hospital discharge. Data analyses were performed from December 2022 to November 2023. Exposure COVID-19 and influenza. Main Outcomes and Measures The primary outcome was major adverse kidney events (MAKE), defined as a composite of mortality and worsened kidney function (estimated glomerular filtration rate [eGFR] decline by ≥25% from discharge eGFR or kidney failure requiring dialysis). Multivariable time-to-event analyses were performed to compare MAKE between individuals with COVID-AKI and those who had AKI associated with other illnesses hospitalized during the same period. For further comparison, this outcome was assessed for a historic cohort of patients with influenza-associated AKI. Results The study cohort included 9624 hospitalized patients (mean [SD] age, 69.0 [15.7] years; 4955 [51.5%] females) with AKI, including 987 patients with COVID-AKI, 276 with influenza-associated AKI, and 8361 with AKI associated with other illnesses (other-AKI). Compared with the other 2 groups, patients with COVID-19-associated AKI were slightly younger in age, had a higher baseline eGFR, worse baseline comorbidity scores, higher markers of illness severity, and longer hospital stay. Compared with the other-AKI group, the COVID-AKI group had lower MAKE (adjusted hazard ratio [aHR], 0.67; 95% CI, 0.59-0.75) due to lower all-cause mortality (aHR, 0.31; 95% CI, 0.24-0.39) and lower rates of worsened kidney function (aHR, 0.78; 95% CI, 0.69-0.88). Conclusions and Relevance The findings of this multicenter cohort study indicate that survivors of hospitalization with COVID-AKI experience lower rates of MAKE, long-term kidney function decline, and mortality compared with patients with AKI associated with other illnesses.
Collapse
Affiliation(s)
- Abinet M. Aklilu
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Sanchit Kumar
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
| | - James Nugent
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
- Section of Nephrology, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - Yu Yamamoto
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
| | - Claudia Coronel-Moreno
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
| | - Bashar Kadhim
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Sophia C. Faulkner
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
| | - Kyle D. O’Connor
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
| | - Farah Yasmin
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
| | - Jason H. Greenberg
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
- Section of Nephrology, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - Dennis G. Moledina
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jeffrey M. Testani
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - F. Perry Wilson
- Clinical and Translational Research Accelerator, Yale University, New Haven, Connecticut
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
10
|
Scigliano NM, Puga TB, Scigliano NM, Williams YK, Boin MA. The Effect of COVID-19 Infection on Clinical Outcomes in Patients Undergoing Surgical Repair of Humerus Fractures. Cureus 2024; 16:e56780. [PMID: 38650794 PMCID: PMC11034398 DOI: 10.7759/cureus.56780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction Limited research exists on the association between coronavirus 2019 (COVID-19) infection and outcomes following surgical fixation for humerus fractures. The objective of this study was to evaluate the effects of COVID-19 on the clinical outcomes of patients undergoing humerus fracture surgery. Methods Approval to utilize insurance claim data from the Change Healthcare dataset was obtained from the Datavant COVID-19 Research Database. Patients older than 55 years old who underwent humerus fracture surgery from April 1, 2020, to March 1, 2022, were included in the analysis. COVID-19 status, comorbidities, and adverse events were identified using the International Classification of Diseases, 10th Revision (ICD-10) diagnostic codes. Propensity score matching with age, sex, and comorbidities was completed to create a 1:10 matched COVID-19-negative cohort. Univariate and multivariate logistic regressions were performed to assess the association of COVID-19 positivity with perioperative adverse events. Results A total of 18,365 patients underwent humerus fracture surgery in this study, of which 132 (0.72%) tested positive for COVID-19. Univariate analysis found that COVID-19-positive patients were at higher risk for myocardial infarction (5.30% vs. 1.74%, p = 0.015) and acute kidney injury (28.79% vs. 12.50%, p < 0.001) when compared to the 1:10 matched COVID-19-negative cohort. In addition, multivariate logistic regression found that COVID-19-positive patients had higher odds of experiencing any adverse event (2.57; 95% CI: 1.69-3.91; p < 0.001) or a minor adverse event (2.44; 95% CI: 1.57-3.79; p < 0.001). Conclusion COVID-19-positive patients have increased odds of experiencing adverse events after undergoing humerus fracture surgery in comparison to a matched COVID-19-negative control. Findings from this study stress the importance of using COVID-19 status as a factor in predicting outcomes following orthopedic surgery in this patient population.
Collapse
Affiliation(s)
| | - Troy B Puga
- Osteopathic Medicine, Kansas City University, Kansas, USA
| | | | - Yale K Williams
- Orthopedic Surgery Residency Program, HCA Research Medical Center, Kansas City University - Graduate Medical Education (GME) Consortium, Kansas, USA
| | - Michael A Boin
- Orthopedic Surgery (Shoulder and Elbow), Orthopedic Health of Kansas City, Kansas, USA
| |
Collapse
|
11
|
Song S, Guo C, Wu R, Zhao H, Li Q, Dou JH, Guo FS, Wei J. Impact of the COVID-19 pandemic on cardiovascular mortality and contrast analysis within subgroups. Front Cardiovasc Med 2024; 11:1279890. [PMID: 38385134 PMCID: PMC10879411 DOI: 10.3389/fcvm.2024.1279890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/18/2024] [Indexed: 02/23/2024] Open
Abstract
Background An increase in deaths has been perceived during the pandemic, which cannot be explained only by COVID-19. The actual number of deaths far exceeds the recorded data on deaths directly related to SARS-CoV-2 infection. Data from early and short-lived pandemic studies show a dramatic shift in cardiovascular mortality. Grounded in the post-pandemic era, macroscopic big data on cardiovascular mortality during the pandemic need to be further reviewed and studied, which is crucial for cardiovascular disease prevention and control. Methods We retrieved and collected data associated with cardiovascular disease mortality from the National Vital Statistic System from the Center for Disease Control and Prevention Wide-Ranging Online Data for Epidemiologic Research (CDC WONDER) platform based on the ICD-10 codes. We applied regression analysis to characterize overall cardiovascular disease mortality trends from 2010 to 2023 and built a time series model to predict mortality for 2020-2023 based on mortality data from 2010 to 2019 in order to affirm the existence of the excess deaths by evaluating observed vs. predicted mortality. We also conducted subgroup analyses by sex, age and race/ethnicity for the purpose of obtaining more specific sociodemographic information. Results All-cause age-standardised mortality rates (ASMRs) for CVD dramatically increased between 2019 and 2021[annual percentage change (APC) 11.27%, p < 0.01], and then decreased in the following 2021-2023(APC: -7.0%, p < 0.01). Subgroup analyses found that the ASMR change was most pronounced in Alaska Indians/Native American people (APC: 16.5% in 2019-2021, -12.5% in 2021-2023, both p < 0.01), Hispanics (APC: 12.1% in 2019-2021, -12.2% in 2021-2023, both p < 0.05) and non-Hispanic Black people (APC:11.8% in 2019-2021, -10.3% in 2021-2023, both p < 0.01)whether during the increasing or declining phase. Similarly, the ASMR change was particularly dramatic for the 25-44 age group (APC:19.8% in 2019-2021, -15.4% in 2021-2023, both p < 0.01) and males (APC: 11.5% in 2019-2021, -7.6% in 2021-2023, both p < 0.01). By the end of 2023, the proportion of COVID-related excess death remained high among the elderly (22.4%), males (42.8%) and Alaska Indians/Native American people(39.7%). In addition, we did not find the presence of excess deaths in the young (25-44) and middle-aged cohort (45-64) in 2023, while excess deaths remained persistent in the elderly. Conclusions All-cause ASMRs for CVD increased notably during the initial two years of the COVID-19 pandemic and then witnessed a decline in 2021-2023. The cohorts (the young, males and minorities) with the steepest rise in mortality decreased at the fastest rate instead. Previous initiatives to promote cardiovascular health were effective, but further research on cardiovascular healthcare for the elderly and racial disparities should be attached to priority considering the presence of sociodemographic differences in CVD death.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jin Wei
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, Xi'an, China
| |
Collapse
|
12
|
Zhang J, Hom K, Zhang C, Nasr M, Gerzanich V, Zhang Y, Tang Q, Xue F, Simard JM, Zhao RY. SARS-CoV-2 ORF3a Protein as a Therapeutic Target against COVID-19 and Long-Term Post-Infection Effects. Pathogens 2024; 13:75. [PMID: 38251382 PMCID: PMC10819734 DOI: 10.3390/pathogens13010075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has posed unparalleled challenges due to its rapid transmission, ability to mutate, high mortality and morbidity, and enduring health complications. Vaccines have exhibited effectiveness, but their efficacy diminishes over time while new variants continue to emerge. Antiviral medications offer a viable alternative, but their success has been inconsistent. Therefore, there remains an ongoing need to identify innovative antiviral drugs for treating COVID-19 and its post-infection complications. The ORF3a (open reading frame 3a) protein found in SARS-CoV-2, represents a promising target for antiviral treatment due to its multifaceted role in viral pathogenesis, cytokine storms, disease severity, and mortality. ORF3a contributes significantly to viral pathogenesis by facilitating viral assembly and release, essential processes in the viral life cycle, while also suppressing the body's antiviral responses, thus aiding viral replication. ORF3a also has been implicated in triggering excessive inflammation, characterized by NF-κB-mediated cytokine production, ultimately leading to apoptotic cell death and tissue damage in the lungs, kidneys, and the central nervous system. Additionally, ORF3a triggers the activation of the NLRP3 inflammasome, inciting a cytokine storm, which is a major contributor to the severity of the disease and subsequent mortality. As with the spike protein, ORF3a also undergoes mutations, and certain mutant variants correlate with heightened disease severity in COVID-19. These mutations may influence viral replication and host cellular inflammatory responses. While establishing a direct link between ORF3a and mortality is difficult, its involvement in promoting inflammation and exacerbating disease severity likely contributes to higher mortality rates in severe COVID-19 cases. This review offers a comprehensive and detailed exploration of ORF3a's potential as an innovative antiviral drug target. Additionally, we outline potential strategies for discovering and developing ORF3a inhibitor drugs to counteract its harmful effects, alleviate tissue damage, and reduce the severity of COVID-19 and its lingering complications.
Collapse
Affiliation(s)
- Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.Z.); (C.Z.)
| | - Kellie Hom
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; (K.H.); (F.X.)
| | - Chenyu Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.Z.); (C.Z.)
| | - Mohamed Nasr
- Drug Development and Clinical Sciences Branch, Division of AIDS, NIAID, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.G.); (J.M.S.)
| | - Yanjin Zhang
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA;
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA;
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; (K.H.); (F.X.)
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.G.); (J.M.S.)
- Research & Development Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.Z.); (C.Z.)
- Research & Development Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
- Department of Microbiology-Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
13
|
Assiri AM, Alamaa T, Elenezi F, Alsagheir A, Alzubaidi L, TIeyjeh I, Alhomod AS, Gaffas EM, Amer SA. Unveiling the Clinical Spectrum of Post-COVID-19 Conditions: Assessment and Recommended Strategies. Cureus 2024; 16:e52827. [PMID: 38406111 PMCID: PMC10884364 DOI: 10.7759/cureus.52827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 02/27/2024] Open
Abstract
SARS-CoV-2 caused the pandemic of the rapidly evolving COVID-19. As of December 6, 2023, there were 765,152,854 COVID-19-recovering cases. Long-term consequences known as "long COVID" and "post-COVID-19 conditions" (PCCs) or "post-acute COVID-19 syndrome" are being reported more frequently in a subset of recovering patients. Systemic, neuropsychiatric, cardio-respiratory, and gastrointestinal symptoms are the most prevalent. The management of PCCs poses unique challenges due to the lack of official guidelines and the complex nature of the illness. This abstract highlights key principles derived from recent reviews and expert recommendations to provide healthcare professionals with a comprehensive approach to manage post-COVID-19 patients. Preventive medicine plays a crucial role in managing PCCs. While no specific medications are available for treatment, preventive measures such as COVID-19 vaccination, adherence to precautionary measures, regular consultations with medical professionals, monitoring symptoms and progress, and seeking information on symptom management are essential to assist patients in their recovery and improve their quality of life. Medical management requires transparent goal-setting and collaborative decision-making based on the patient's symptoms, comorbidities, and treatment objectives. Treatment plans for post-COVID-19 patients should focus on patient education, using registries and calendars to track symptoms and triggers, providing support and reassurance, and offering holistic support through peer networks and supportive psychotherapy techniques. Symptomatic and rehabilitative care, including well-established symptom management techniques, physical rehabilitation programs, and addressing mental health and well-being, are vital components of post-COVID-19 management. Lifestyle factors such as stress reduction, nutrition, and sleep should be incorporated into managing underlying medical conditions in post-COVID-19 patients. Regular follow-up visits and referrals to specialists are recommended to monitor the patient's progress and address specific organ system involvement or additional care needs. In summary, for the effective management of PCCs, a holistic approach should include preventive measures, patient education, supportive psychotherapy, symptomatic and rehabilitative care, medical management, counseling on lifestyle elements, and appropriate follow-up plans. However, it is crucial to stay updated with evolving guidelines and recommendations from healthcare authorities to provide the most effective and evidence-based care to post-COVID-19 patients.
Collapse
Affiliation(s)
| | - Tareef Alamaa
- Therapeutic Services, Saudi Ministry of Health, Riyadh, SAU
| | - Faisal Elenezi
- Assistant Agency for Hospital Affairs, Saudi Ministry of Health, Riyadh, SAU
| | - Aeshah Alsagheir
- Health Quality Index Measuring, Saudi Ministry of Health, Riyadh, SAU
| | - Lamya Alzubaidi
- Assistant Agency for Hospital Affairs, Saudi Ministry of Health, Riyadh, SAU
| | - Imad TIeyjeh
- Infectious Diseases, Mayo Clinic, Rochester, USA
- Infectious Diseases, King Fahad Medical City, Riyadh, SAU
| | | | - Eisha M Gaffas
- Mental Health and Social Services, Saudi Ministry of Health, Riyadh, SAU
| | - Samar A Amer
- Public Health and Community Medicine, Zagazig University, Zagazig, EGY
- General Administration of Health Programs and Non-communicable Diseases, Saudi Ministry of Health, Riyadh, SAU
| |
Collapse
|
14
|
Zhang Y, Zhao Y, Wang J, Zheng X, Xu D, Lv J, Yang L. Long-term renal outcomes of patients with COVID-19: a meta-analysis of observational studies. J Nephrol 2023; 36:2441-2456. [PMID: 37787893 DOI: 10.1007/s40620-023-01731-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/03/2023] [Indexed: 10/04/2023]
Abstract
BACKGROUND Kidney involvement is common in hospitalized coronavirus disease 2019 (COVID-19) patients during the acute phase, little is known about the long-term impact of COVID-19 on the kidney. METHODS This is a systematic review and meta-analysis on long-term renal outcomes among COVID-19 patients. We carried out a systematic literature search in PUBMED, EMBASE, SCOPUS, and Cochrane COVID-19 study register and performed the random-effects meta-analysis of rates. The search was last updated on November 23, 2022. RESULTS The study included 12 moderate to high-quality cohort studies involving 6976 patients with COVID-19-associated acute kidney injury and 5223 COVID-19 patients without acute kidney injury. The summarized long-term renal non-recovery rate, dialysis-dependent rate, and complete recovery rate among patients with COVID-19-associated AKI was 22% (12-33%), 6% (2-12%), and 63% (44-81%) during a follow-up of 90-326.5 days. Heterogeneity could be explained by differences in the prevalence of chronic kidney disease and proportion of acute kidney injury requiring renal replacement therapy using meta-regression; patients with more comorbidities or higher renal replacement therapy rate had higher non-recovery rates. The summarized long-term kidney function decrease rate among patients without acute kidney injury was 22% (3-51%) in 90-199 days, with heterogeneity partially explained by severity of infection. CONCLUSION Patients with more comorbidities tend to have a higher renal non recovery rate after COVID-19-associated acute kidney injury; for COVID-19 patients without acute kidney injury, decrease in kidney function may occur during long-term follow-up. Regular evaluation of kidney function during the post-COVID-19 follow-up among high-risk patients may be necessary.
Collapse
Affiliation(s)
- Yuhui Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Youlu Zhao
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Jinwei Wang
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Xizi Zheng
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Damin Xu
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Jicheng Lv
- Renal Division, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health, Beijing, China
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | - Li Yang
- Renal Division, Peking University First Hospital, Beijing, China.
- Institute of Nephrology, Peking University, Beijing, China.
- Key Laboratory of Renal Disease, Ministry of Health, Beijing, China.
- Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China.
- Research Units of Diagnosis and Treatment of lmmune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
15
|
Rai V. COVID-19 and Kidney: The Importance of Follow-Up and Long-Term Screening. Life (Basel) 2023; 13:2137. [PMID: 38004277 PMCID: PMC10672056 DOI: 10.3390/life13112137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/21/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
Renal involvement and kidney injury are common in COVID-19 patients, and the symptoms are more severe if the patient already has renal impairment. Renal involvement in COVID-19 is multifactorial, and the renal tubule is mainly affected, along with podocyte injury during SARS-CoV-2 infection. Inflammation, complement activation, hypercoagulation, and crosstalk between the kidney and lungs, brain, and heart are contributory factors. Kidney injury during the acute phase, termed acute kidney injury (AKI), may proceed to chronic kidney disease if the patient is discharged with renal impairment. Both AKI and chronic kidney disease (CKD) increase mortality in COVID-19 patients. Further, COVID-19 infection in patients suffering from CKD is more severe and increases the mortality rate. Thus, it is important to address both categories of patients, either developing AKI or CKD after COVID-19 or previously having CKD, with proper management and treatment. This review discusses the pathophysiology involved in AKI and CKD in COVID-19 infection, followed by management and treatment of AKI and CKD. This is followed by a discussion of the importance of screening and treatment of CKD patients infected with COVID-19 and future perspectives to improve treatment in such patients.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
16
|
Zechendorf E, Beckers C, Frank N, Kraemer S, Neu C, Breuer T, Dreher M, Dahl E, Marx G, Martin L, Simon TP. A Potential Association between Ribonuclease 1 Dynamics in the Blood and the Outcome in COVID-19 Patients. Int J Mol Sci 2023; 24:12428. [PMID: 37569802 PMCID: PMC10419077 DOI: 10.3390/ijms241512428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
The COVID-19 pandemic caused by the new SARS-CoV-2 coronavirus is the most recent and well-known outbreak of a coronavirus. RNase 1 is a small endogenous antimicrobial polypeptide that possesses antiviral activity against viral diseases. In this study, we investigated a potential association between ribonuclease 1 and the outcome in COVID-19 patients and the impact of increased and decreased RNase 1 levels serum during the course of the disease. Therefore, two patient populations, Cohort A (n = 35) and B (n = 80), were subclassified into two groups, in which the RNase 1 concentration increased or decreased from time point one to time point two. We show that the RNase 1 serum levels significantly increased in the increasing group of both cohorts (p = 0.0171; p < 0.0001). We detect that patients in the increasing group who died had significantly higher RNase 1 serum levels at both time points in Cohort A (p = 0.0170; p = 0.0393) and Cohort B (p = 0.0253; p = 0.0034) than patients who survived. Additionally, we measured a significant correlation of RNase 1 serum levels with serum creatinine as well as creatinine clearance in the increasing and decreasing group at both time points of Cohort A. Based on these results, there is now good evidence that RNase 1 may play a role in renal dysfunction associated with ICU COVID-19 patients and that increasing RNase 1 serum level may be a potential biomarker to predict outcome in COVID-19 patients.
Collapse
Affiliation(s)
- Elisabeth Zechendorf
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.-P.S.)
| | - Christian Beckers
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.-P.S.)
| | - Nadine Frank
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.-P.S.)
| | - Sandra Kraemer
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.-P.S.)
| | - Carolina Neu
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.-P.S.)
| | - Thomas Breuer
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.-P.S.)
| | - Michael Dreher
- Department of Pneumology and Intensive Care Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Edgar Dahl
- RWTH Centralized Biomaterial Bank (RWTH cBMB) at the Institute of Pathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Gernot Marx
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.-P.S.)
| | - Lukas Martin
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.-P.S.)
| | - Tim-Philipp Simon
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.-P.S.)
| |
Collapse
|
17
|
Qin F, Wang H, Li M, Zhuo S, Liu W. Drug-drug interaction of Nirmatrelvir/ritonavir and tacrolimus: A potential risk disproportionality analysis of nephrotoxicity from COVID-19 reports in FAERS. Expert Opin Drug Saf 2023; 22:1321-1327. [PMID: 37477905 DOI: 10.1080/14740338.2023.2239156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/22/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Nirmatrelvir/ritonavir is a new oral antiviral agent for COVID-19, and tacrolimus is a widely used immunosuppressant. Drug-drug interaction between Nirmatrelvir/ritonavir and tacrolimus is expected. However, information regarding the drug-drug interaction in a real-world setting is limited. We aim to evaluate drug-drug interaction between tacrolimus and Nirmatrelvir/ritonavir and perform a disproportionality analysis to assess the potential risk of nephrotoxicity due to their combination for COVID-19 treatment based on the FAERS database. RESEARCH DESIGN AND METHODS Disproportionality analysis was performed using the reporting odds ratio (ROR) method, and subset analysis was conducted based on the background of COVID-19 drugs combined with tacrolimus more than 10 times. RESULTS In disproportionality analysis, combination of Nirmatrelvir/ritonavir and tacrolimus was significantly associated with acute kidney injury (41.13%), serum creatinine increased (14.18%), renal failure (2.84%), and renal impairment (2.84%). These positive signals of acute kidney injury and serum creatinine increased still strongly retained in subset analysis. No similar positive signals were detected in Nirmatrelvir/ritonavir-single group. Only in Cilgavimab/Tixagevimab-tacrolimus group and Remdesivir-tacrolimus group, acute kidney injury was recognized as weakly positive signals and disappeared in subset analysis. CONCLUSIONS The study results show significant drug-drug interaction between Nirmatrelvir/ritonavir and tacrolimus and confirm that their combination for COVID-19 treatment greatly increases risk of acute kidney injury.
Collapse
Affiliation(s)
- Fuhong Qin
- Department of Pharmacy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huiling Wang
- School of Pharmaceutical Sciences, Zhengzhou Railway Vocational and Technical College, Zhengzhou, China
| | - Meng Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shengnan Zhuo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wei Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
Yazdanparast S, Bakhtiyaridovvombaygi M, Mikanik F, Ahmadi R, Ghorbani M, Mansoorian MR, Mansoorian M, Chegni H, Moshari J, Gharehbaghian A. Spotlight on contributory role of host immunogenetic profiling in SARS-CoV-2 infection: Susceptibility, severity, mortality, and vaccine effectiveness. Life Sci 2023:121907. [PMID: 37394094 DOI: 10.1016/j.lfs.2023.121907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND The SARS-CoV-2 virus has spread continuously worldwide, characterized by various clinical symptoms. The immune system responds to SARS-CoV-2 infection by producing Abs and secreting cytokines. Recently, numerous studies have highlighted that immunogenetic factors perform a putative role in COVID-19 pathogenesis and implicate vaccination effectiveness. AIM This review summarizes the relevant articles and evaluates the significance of mutation and polymorphism in immune-related genes regarding susceptibility, severity, mortality, and vaccination effectiveness of COVID-19. Furthermore, the correlation between host immunogenetic and SARS-CoV-2 reinfection is discussed. METHOD A comprehensive search was conducted to identify relevant articles using five databases until January 2023, which resulted in 105 total articles. KEY FINDINGS Taken to gather this review summarized that: (a) there is a plausible correlation between immune-related genes and COVID-19 outcomes, (b) the HLAs, cytokines, chemokines, and other immune-related genes expression profiles can be a prognostic factor in COVID-19-infected patients, and (c) polymorphisms in immune-related genes have been associated with the effectiveness of vaccination. SIGNIFICANCE Regarding the importance of mutation and polymorphisms in immune-related genes in COVID-19 outcomes, modulating candidate genes is expected to help clinical decisions, patient outcomes management, and innovative therapeutic approach development. In addition, the manipulation of host immunogenetics is hypothesized to induce more robust cellular and humoral immune responses, effectively increase the efficacy of vaccines, and subsequently reduce the incidence rates of reinfection-associated COVID-19.
Collapse
Affiliation(s)
- Somayeh Yazdanparast
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Bakhtiyaridovvombaygi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mikanik
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Ahmadi
- Department of Infectious Diseases, School of Medicine, Infectious Diseases Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Mohammad Ghorbani
- Laboratory Hematology and Transfusion Medicine, Department of Pathology, Faculty Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| | | | - Mozhgan Mansoorian
- Nursing Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Hamid Chegni
- Department of Immunology, School of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalil Moshari
- School of Medicine, Gonabad University of Medical Science, Gonabad, Iran
| | - Ahmad Gharehbaghian
- Department of Hematology and Blood Bank, School of Allied Medical Science, Shahid Beheshti University of Medical Science, Tehran, Iran; Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Zhou H, Xu M, Hu P, Li Y, Ren C, Li M, Pan Y, Wang S, Liu X. Identifying hub genes and common biological pathways between COVID-19 and benign prostatic hyperplasia by machine learning algorithms. Front Immunol 2023; 14:1172724. [PMID: 37426635 PMCID: PMC10328422 DOI: 10.3389/fimmu.2023.1172724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023] Open
Abstract
Background COVID-19, a serious respiratory disease that has the potential to affect numerous organs, is a serious threat to the health of people around the world. The objective of this article is to investigate the potential biological targets and mechanisms by which SARS-CoV-2 affects benign prostatic hyperplasia (BPH) and related symptoms. Methods We downloaded the COVID-19 datasets (GSE157103 and GSE166253) and the BPH datasets (GSE7307 and GSE132714) from the Gene Expression Omnibus (GEO) database. In GSE157103 and GSE7307, differentially expressed genes (DEGs) were found using the "Limma" package, and the intersection was utilized to obtain common DEGs. Further analyses followed, including those using Protein-Protein Interaction (PPI), Gene Ontology (GO) function enrichment analysis, and the Kyoto Encyclopedia of Genes and Genomes (KEGG). Potential hub genes were screened using three machine learning methods, and they were later verified using GSE132714 and GSE166253. The CIBERSORT analysis and the identification of transcription factors, miRNAs, and drugs as candidates were among the subsequent analyses. Results We identified 97 common DEGs from GSE157103 and GSE7307. According to the GO and KEGG analyses, the primary gene enrichment pathways were immune-related pathways. Machine learning methods were used to identify five hub genes (BIRC5, DNAJC4, DTL, LILRB2, and NDC80). They had good diagnostic properties in the training sets and were validated in the validation sets. According to CIBERSORT analysis, hub genes were closely related to CD4 memory activated of T cells, T cells regulatory and NK cells activated. The top 10 drug candidates (lucanthone, phytoestrogens, etoposide, dasatinib, piroxicam, pyrvinium, rapamycin, niclosamide, genistein, and testosterone) will also be evaluated by the P value, which is expected to be helpful for the treatment of COVID-19-infected patients with BPH. Conclusion Our findings reveal common signaling pathways, possible biological targets, and promising small molecule drugs for BPH and COVID-19. This is crucial to understand the potential common pathogenic and susceptibility pathways between them.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mingming Xu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Hu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuezheng Li
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Congzhe Ren
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Muwei Li
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang Pan
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shangren Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
20
|
Tolmacheva AS, Onvumere MK, Sedykh SE, Timofeeva AM, Nevinsky GA. Catalase Activity of IgGs of Patients Infected with SARS-CoV-2. Int J Mol Sci 2023; 24:10081. [PMID: 37373231 DOI: 10.3390/ijms241210081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/06/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Coronavirus disease (COVID-19), caused by the SARS-CoV-2 coronavirus, leads to various manifestations of the post-COVID syndrome, including diabetes, heart and kidney disease, thrombosis, neurological and autoimmune diseases and, therefore, remains, so far, a significant public health problem. In addition, SARS-CoV-2 infection can lead to the hyperproduction of reactive oxygen species (ROS), causing adverse effects on oxygen transfer efficiency, iron homeostasis, and erythrocytes deformation, contributing to thrombus formation. In this work, the relative catalase activity of the serum IgGs of patients recovered from COVID-19, healthy volunteers vaccinated with Sputnik V, vaccinated with Sputnik V after recovering from COVID-19, and conditionally healthy donors were analyzed for the first time. Previous reports show that along with canonical antioxidant enzymes, the antibodies of mammals with superoxide dismutase, peroxidase, and catalase activities are involved in controlling reactive oxygen species levels. We here show that the IgGs from patients who recovered from COVID-19 had the highest catalase activity, and this was statistically significantly higher each compared to the healthy donors (1.9-fold), healthy volunteers vaccinated with Sputnik V (1.4-fold), and patients vaccinated after recovering from COVID-19 (2.1-fold). These data indicate that COVID-19 infection may stimulate the production of antibodies that degrade hydrogen peroxide, which is harmful at elevated concentrations.
Collapse
Affiliation(s)
- Anna S Tolmacheva
- Institute of Chemical Biology and Fundamental Medicine, SB of the RAS, 630090 Novosibirsk, Russia
| | - Margarita K Onvumere
- Institute of Chemical Biology and Fundamental Medicine, SB of the RAS, 630090 Novosibirsk, Russia
| | - Sergey E Sedykh
- Institute of Chemical Biology and Fundamental Medicine, SB of the RAS, 630090 Novosibirsk, Russia
| | - Anna M Timofeeva
- Institute of Chemical Biology and Fundamental Medicine, SB of the RAS, 630090 Novosibirsk, Russia
| | - Georgy A Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, SB of the RAS, 630090 Novosibirsk, Russia
| |
Collapse
|
21
|
Montani D, Savale L, Noel N, Meyrignac O, Colle R, Gasnier M, Corruble E, Beurnier A, Jutant EM, Pham T, Lecoq AL, Papon JF, Figuereido S, Harrois A, Humbert M, Monnet X. [Post-COVID-19 syndrome]. BULLETIN DE L'ACADEMIE NATIONALE DE MEDECINE 2023; 207:812-820. [PMID: 37292432 PMCID: PMC10126882 DOI: 10.1016/j.banm.2023.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/17/2023] [Indexed: 06/10/2023]
Abstract
In the aftermath of acute infection with the severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2), a large number of symptoms persist or appear, constituting a real syndrome called "long COVID-19" or "post-COVID- 19" or "post-acute COVID-19 syndrome". Its incidence is very high, half of patients showing at least one symptom at 4-6 months after Coronarovirus infectious disease 2019 (COVID-19). They can affect many organs. The most common symptom is persistent fatigue, similar to that seen after other viral infections. Radiological pulmonary sequelae are relatively rare and not extensive. On the other hand, functional respiratory symptoms, primarily dyspnoea, are much more frequent. Dysfunctional breathing is a significant cause of dyspnoea. Cognitive disorders and psychological symptoms are also very common, with anxiety, depression and post-traumatic stress symptoms being widely described. On the other hand, cardiac, endocrine, cutaneous, digestive or renal sequelae are rarer. The symptoms generally improve after several months, even if their prevalence at two years remains significant. Most of the symptoms are favored by the severity of the initial illness, and the psychic symptoms by the female sex. The pathophysiology of most symptoms is poorly understood. The influence of the treatments used in the acute phase is also important. Vaccination, on the other hand, seems to reduce their incidence. The sheer number of affected patients makes long-term COVID-19 syndrome a public health challenge.
Collapse
Affiliation(s)
- David Montani
- Université Paris-Saclay, AP-HP, service de pneumologie et soins intensifs respiratoires, hôpital de Bicêtre, DMU 5 Thorinno, Inserm UMR_S999, Le Kremlin-Bicêtre, France
| | - Laurent Savale
- Université Paris-Saclay, AP-HP, service de pneumologie et soins intensifs respiratoires, hôpital de Bicêtre, DMU 5 Thorinno, Inserm UMR_S999, Le Kremlin-Bicêtre, France
| | - Nicolas Noel
- Université Paris-Saclay, AP-HP, service de médecine interne et immunologie clinique, hôpital de Bicêtre, DMU 7 endocrinologie-immunités-inflammations-cancer-urgences, Le Kremlin-Bicêtre, France
| | - Olivier Meyrignac
- Université Paris-Saclay, AP-HP, service de radiologie diagnostique et interventionnelle, Hôpital de Bicêtre, DMU 14 Smart Imaging, BioMaps, Le Kremlin-Bicêtre, France
| | - Romain Colle
- Université Paris-Saclay, AP-HP, service de psychiatrie, hôpital de Bicêtre, DMU 11 psychiatrie, santé mentale, addictologie et nutrition, équipe MOODS, Inserm U1178, centre de recherche en épidémiologie et santé des populations (CESP), Le Kremlin-Bicêtre, France
| | - Matthieu Gasnier
- Université Paris-Saclay, AP-HP, service de psychiatrie, hôpital de Bicêtre, DMU 11 psychiatrie, santé mentale, addictologie et nutrition, équipe MOODS, Inserm U1178, centre de recherche en épidémiologie et santé des populations (CESP), Le Kremlin-Bicêtre, France
| | - Emmanuelle Corruble
- Université Paris-Saclay, AP-HP, service de psychiatrie, hôpital de Bicêtre, DMU 11 psychiatrie, santé mentale, addictologie et nutrition, équipe MOODS, Inserm U1178, centre de recherche en épidémiologie et santé des populations (CESP), Le Kremlin-Bicêtre, France
| | - Antoine Beurnier
- Université Paris-Saclay, AP-HP, service de pneumologie et soins intensifs respiratoires, hôpital de Bicêtre, DMU 5 Thorinno, Inserm UMR_S999, Le Kremlin-Bicêtre, France
| | - Etienne-Marie Jutant
- Université Paris-Saclay, AP-HP, service de pneumologie et soins intensifs respiratoires, hôpital de Bicêtre, DMU 5 Thorinno, Inserm UMR_S999, Le Kremlin-Bicêtre, France
- Université de Poitiers, CHU de Poitiers, service de pneumologie, Inserm CIC 1402 Axe Is-ALIVE, Poitiers, France
| | - Tai Pham
- Université Paris-Saclay, AP-HP, service de médecine intensive-réanimation, hôpital de Bicêtre, DMU 4 CORREVE maladies du cœur et des vaisseaux, Inserm UMR_S999, FHU Sepsis, CARMAS, 78, rue du Général-Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Anne-Lise Lecoq
- Université Paris-Saclay, AP-HP, centre de recherche clinique Paris-Saclay, DMU 13 santé publique, information médicale, appui à la recherche clinique, Inserm U1018, centre de recherche en épidémiologie et santé des populations (CESP), Le Kremlin-Bicêtre, France
| | - Jean-François Papon
- Université Paris-Saclay, AP-HP, service d'ORL et de chirurgie cervico-faciale, DMU 9 neurosciences, Inserm, U955, E13, CNRS ERL7000, Le Kremlin-Bicêtre, France
| | - Samy Figuereido
- Université Paris-Saclay, AP-HP, service d'anesthésie-réanimation et médecine péri-opératoire, Hôpital de Bicêtre, DMU 12 anesthésie, réanimation, douleur, Le Kremlin-Bicêtre, France
| | - Anatole Harrois
- Université Paris-Saclay, AP-HP, service de réanimation chirurgicale, hôpital de Bicêtre, DMU 12 anesthésie, réanimation, douleur, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- Université Paris-Saclay, AP-HP, service de pneumologie et soins intensifs respiratoires, hôpital de Bicêtre, DMU 5 Thorinno, Inserm UMR_S999, Le Kremlin-Bicêtre, France
| | - Xavier Monnet
- Université Paris-Saclay, AP-HP, service de médecine intensive-réanimation, hôpital de Bicêtre, DMU 4 CORREVE maladies du cœur et des vaisseaux, Inserm UMR_S999, FHU Sepsis, CARMAS, 78, rue du Général-Leclerc, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
22
|
Marshall GD. The pathophysiology of postacute sequelae of COVID-19 (PASC): Possible role for persistent inflammation. Asia Pac Allergy 2023; 13:77-84. [PMID: 37388814 PMCID: PMC10287107 DOI: 10.5415/apallergy.0000000000000106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 07/01/2023] Open
Abstract
As the SARS-CoV-2-induced pandemic wanes, a substantial number of patients with acute Corona Virus-induced disease (COVID-19 continue to have symptoms for a prolonged time after initial infection. These patients are said to have postacute sequelae of COVID (PASC) or "long COVID". The underlying pathophysiology of this syndrome is poorly understood and likely quite heterogeneous. The role of persistent, possibly deviant inflammation as a major factor in comorbidity is suspected. Objective To review data that address the relative importance of inflammation in the pathophysiology spectrum of PASC and to address how this would impact diagnosis and approach to therapy in patients identified as having such inflammatory abnormalities. Methods A review of public databases, including PubMed, MeSH, NLM catalog, and clinical trial databases such as clinicaltrials.gov. Results The literature supports a prominent role for various forms and types of inflammation in the pathophysiologic spectrum of PASC. Such inflammation can be persistent ant CoV-2-specific responses, new onset autoimmune responses, or a loss of normal immunoregulation resulting in widespread, sustained inflammatory pathologies that can affect both broad constitutional symptoms (such as fatigue, neurocognitive dysfunction, and anxiety/depression) and organ-specific dysfunction and/or failure. Conclusions PASC is a significant clinical entity with similarities to and differences from other postviral syndromes. Significant research efforts are ongoing to better understand specific aberrant inflammatory pathways present in individual patients for the purpose of developing and implementing effective therapies and ultimately prophylaxis strategies to prevent the progression of COVID-19 as well as likely future viral illnesses and pandemics.
Collapse
Affiliation(s)
- Gailen D. Marshall
- Department of Medicine, The University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
23
|
Murali R, Wanjari UR, Mukherjee AG, Gopalakrishnan AV, Kannampuzha S, Namachivayam A, Madhyastha H, Renu K, Ganesan R. Crosstalk between COVID-19 Infection and Kidney Diseases: A Review on the Metabolomic Approaches. Vaccines (Basel) 2023; 11:vaccines11020489. [PMID: 36851366 PMCID: PMC9959335 DOI: 10.3390/vaccines11020489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19, a respiratory disorder. Various organ injuries have been reported in response to this virus, including kidney injury and, in particular, kidney tubular injury. It has been discovered that infection with the virus does not only cause new kidney disease but also increases treatment difficulty and mortality rates in people with kidney diseases. In individuals hospitalized with COVID-19, urinary metabolites from several metabolic pathways are used to distinguish between patients with acute kidney injury (AKI) and those without. This review summarizes the pathogenesis, pathophysiology, treatment strategies, and role of metabolomics in relation to AKI in COVID-19 patients. Metabolomics is likely to play a greater role in predicting outcomes for patients with kidney disease and COVID-19 with varying levels of severity in the near future as data on metabolic profiles expand rapidly. Here, we also discuss the correlation between COVID-19 and kidney diseases and the available metabolomics approaches.
Collapse
Affiliation(s)
- Reshma Murali
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
- Correspondence: (A.V.G.); (R.G.)
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Arunraj Namachivayam
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Kaviyarasi Renu
- Center of Molecular Medicine and Diagnostics (COMMAND), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Correspondence: (A.V.G.); (R.G.)
| |
Collapse
|
24
|
Zavori L, Molnar T, Varnai R, Kanizsai A, Nagy L, Vadkerti B, Szirmay B, Schwarcz A, Csecsei P. Cystatin-c May Indicate Subclinical Renal Involvement, While Orosomucoid Is Associated with Fatigue in Patients with Long-COVID Syndrome. J Pers Med 2023; 13:371. [PMID: 36836605 PMCID: PMC9958557 DOI: 10.3390/jpm13020371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Long-COVID syndrome is associated with high healthcare costs, but its pathophysiology is not yet fully understood. Inflammation, renal impairment or disturbance of the NO system emerge as potential pathogenetic factors. We aimed to investigate the relationship between symptoms of long-COVID syndrome and serum levels of cystatin-c (CYSC), orosomucoid (ORM), l-arginine, symmetric dimethylarginine (SDMA) and asymmetric dimethylarginine (ADMA). A total of 114 patients suffering from long-COVID syndrome were included in this observational cohort study. We found that serum CYSC was independently associated with the anti-spike immunoglobulin (S-Ig) serum level (OR: 5.377, 95% CI: 1.822-12.361; p = 0.02), while serum ORM (OR: 9.670 (95% CI: 1.34-9.93; p = 0.025) independently predicted fatigue in patients with long-COVID syndrome, both measured at baseline visit. Additionally, the serum CYSC concentrations measured at the baseline visit showed a positive correlation with the serum SDMA levels. The severity of abdominal and muscle pain indicated by patients at the baseline visit showed a negative correlation with the serum level of L-arginine. In summary, serum CYSC may indicate subclinical renal impairment, while serum ORM is associated with fatigue in long-COVID syndrome. The potential role of l-arginine in alleviating pain requires further studies.
Collapse
Affiliation(s)
- Laszlo Zavori
- Salisbury NHS Foundation Trust, Salisbury SP2 8BJ, UK
| | - Tihamer Molnar
- Department of Anaesthesiology and Intensive Care, Medical School, University of Pecs, 7624 Pecs, Hungary
| | - Reka Varnai
- Department of Primary Health Care, Medical School, University of Pecs, 7624 Pecs, Hungary
| | - Andrea Kanizsai
- Department of Dentistry, Medical School, Pecs, University of Pecs, 7624 Pecs, Hungary
| | - Lajos Nagy
- Department of Applied Chemistry, University of Debrecen, 4032 Debrecen, Hungary
| | - Bence Vadkerti
- Department of Applied Chemistry, University of Debrecen, 4032 Debrecen, Hungary
| | - Balazs Szirmay
- Department of Laboratory Medicine, Medical School, University of Pecs, 7624 Pecs, Hungary
| | - Attila Schwarcz
- Department of Neurosurgery, Medical School, University of Pecs, 7624 Pecs, Hungary
| | - Peter Csecsei
- Department of Neurosurgery, Medical School, University of Pecs, 7624 Pecs, Hungary
| |
Collapse
|
25
|
Lutchmansingh DD, Higuero Sevilla JP, Possick JD, Gulati M. "Long Haulers". Semin Respir Crit Care Med 2023; 44:130-142. [PMID: 36646091 DOI: 10.1055/s-0042-1759568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Post-COVID conditions continue to afflict patients long after acute severe acute respiratory syndrome-coronavirus-2 (SARS CoV-2) infection. Over 50 symptoms across multiple organ systems have been reported, with pulmonary, cardiovascular, and neuropsychiatric sequelae occurring most frequently. Multiple terms have been used to describe post-COVID conditions including long COVID, long-haul COVID, postacute coronavirus disease 2019 (COVID-19), postacute sequelae of SARS-CoV-2 infection, long-term effects of COVID, and chronic COVID-19; however, standardized assessments and treatment algorithms for patients have generally been lacking. This review discusses the epidemiology and risk factors for post-COVID conditions and provides a general overview of the diagnostic assessment and treatment of specific manifestations. Data derived from the multitude of observational studies and scientific investigations into pathogenesis are providing a clearer understanding of the distinct phenotypes of post-COVID conditions. Insight gained from these studies and ongoing interventional trials continues to lead to the development of clinical protocols directed toward improving COVID-19 survivors' quality of life and preventing or reducing long-term morbidity.
Collapse
Affiliation(s)
- Denyse D Lutchmansingh
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jean Paul Higuero Sevilla
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jennifer D Possick
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Mridu Gulati
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
26
|
McColl ER, Croyle MA, Zamboni WC, Honer WG, Heise M, Piquette-Miller M, Goralski KB. COVID-19 Vaccines and the Virus: Impact on Drug Metabolism and Pharmacokinetics. Drug Metab Dispos 2023; 51:130-141. [PMID: 36273826 PMCID: PMC11022893 DOI: 10.1124/dmd.122.000934] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/07/2022] [Accepted: 09/30/2022] [Indexed: 01/08/2023] Open
Abstract
This article reports on an American Society of Pharmacology and Therapeutics, Division of Drug Metabolism and Disposition symposium held at Experimental Biology on April 2, 2022, in Philadelphia. As of July 2022, over 500 million people have been infected with SARS-CoV-2 (the virus causing COVID-19) and over 12 billion vaccine doses have been administered. Clinically significant interactions between viral infections and hepatic drug metabolism were first recognized over 40 years ago during a cluster of pediatric theophylline toxicity cases attributed to reduced hepatic drug metabolism amid an influenza B outbreak. Today, a substantive body of research supports that the activated innate immune response generally decreases hepatic cytochrome P450 activity. The interactions extend to drug transporters and other organs and have the potential to impact drug absorption, distribution, metabolism, and excretion (ADME). Based on this knowledge, altered ADME is predicted with SARS-CoV-2 infection or vaccination. The report begins with a clinical case exploring the possibility of SARS-CoV-2 vaccination increasing clozapine levels. This is followed by discussions of how SARS-CoV-2 infection or vaccines alter the metabolism and disposition of complex drugs, such as nanoparticles and biologics and small molecule therapies. The review concludes with a discussion of the effects of viral infections on placental amino acid transport and their potential to impact fetal development. The session improved our understanding of the impact of emerging viral infections and vaccine technologies on drug metabolism and disposition, which will help mitigate drug toxicity and improve drug and vaccine safety and effectiveness. SIGNIFICANCE STATEMENT: Altered pharmacokinetics of small molecule and complex molecule drugs and fetal brain distribution of amino acids following SARS-CoV-2 infection or immunization are possible. The proposed mechanisms involve decreased liver cytochrome P450 metabolism of small molecules, enhanced innate immune system metabolism of complex molecules, and altered placental and fetal blood-brain barrier amino acid transport, respectively. Future research is needed to understand the effects of these interactions on adverse drug responses, drug and vaccine safety, and effectiveness and fetal neurodevelopment.
Collapse
Affiliation(s)
- Eliza R McColl
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Maria A Croyle
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - William C Zamboni
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - William G Honer
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Mark Heise
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Micheline Piquette-Miller
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Kerry B Goralski
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| |
Collapse
|
27
|
McAlister FA, Parikh H, Lee DS, Wijeysundera HC. Health Care Implications of the COVID-19 Pandemic for the Cardiovascular Practitioner. Can J Cardiol 2022:S0828-282X(22)01051-0. [PMID: 36481398 PMCID: PMC9721374 DOI: 10.1016/j.cjca.2022.11.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
There has been substantial excess morbidity and mortality during the COVID-19 pandemic, not all of which was directly attributable to SARS-CoV-2 infection, and many non-COVID-19 deaths were cardiovascular. The indirect effects of the pandemic have been profound, resulting in a substantial increase in the burden of cardiovascular disease and cardiovascular risk factors, both in individuals who survived SARS-CoV-2 infection and in people never infected. In this report, we review the direct effect of SARS-CoV-2 infection on cardiovascular and cardiometabolic disease burden in COVID-19 survivors as well as the indirect effects of the COVID-19 pandemic on the cardiovascular health of people who were never infected with SARS-CoV-2. We also examine the pandemic effects on health care systems and particularly the care deficits caused (or exacerbated) by health care delayed or foregone during the COVID-19 pandemic. We review the consequences of: (1) deferred/delayed acute care for urgent conditions; (2) the shift to virtual provision of outpatient care; (3) shortages of drugs and devices, and reduced access to: (4) diagnostic testing, (5) cardiac rehabilitation, and (6) homecare services. We discuss the broader implications of the COVID-19 pandemic for cardiovascular health and cardiovascular practitioners as we move forward into the next phase of the pandemic.
Collapse
Affiliation(s)
- Finlay A. McAlister
- The Division of General Internal Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada,The Alberta Strategy for Patient Oriented Research Support Unit, Edmonton, Alberta, Canada,Corresponding author: Dr Finlay A. McAlister, 5-134C Clinical Sciences Building, University of Alberta, 11350 83 Avenue, Edmonton, Alberta T6G 2G3, Canada. Tel.: +1-780-492-9824; fax: +1-780-492-7277
| | - Harsh Parikh
- Peter Munk Cardiac Center, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Douglas S. Lee
- Peter Munk Cardiac Center, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada,ICES (formerly Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada
| | - Harindra C. Wijeysundera
- ICES (formerly Institute for Clinical Evaluative Sciences), Toronto, Ontario, Canada,Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada,Schulich Heart Program, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Cellular miR-6741-5p as a Prognostic Biomarker Predicting Length of Hospital Stay among COVID-19 Patients. Viruses 2022; 14:v14122681. [PMID: 36560686 PMCID: PMC9781286 DOI: 10.3390/v14122681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/20/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Wide variability exists with host response to SARS-CoV-2 infection among individuals. Circulatory micro RNAs (miRNAs) are being recognized as promising biomarkers for complex traits, including viral pathogenesis. We hypothesized that circulatory miRNAs at 48 h post hospitalization may predict the length of stay (LOS) and prognosis of COVID-19 patients. Plasma miRNA levels were compared between three groups: (i) healthy volunteers (C); (ii) COVID-19 patients treated with remdesivir (an antiviral) plus dexamethasone (a glucocorticoid) (with or without baricitinib, a Janus kinase inhibitor) on the day of hospitalization (I); and COVID-19 patients at 48 h post treatment (T). Results showed that circulatory miR-6741-5p expression levels were significantly different between groups C and I (p < 0.0000001); I and T (p < 0.0000001); and C and T (p = 0.001). Our ANOVA model estimated that all patients with less than 12.42 Log2 CPM had a short LOS, or a good prognosis, whereas all patients with over 12.42 Log2 CPM had a long LOS, or a poor prognosis. In sum, we show that circulatory miR-6741-5p may serve as a prognostic biomarker effectively predicting mortality risk and LOS of hospitalized COVID-19 patients.
Collapse
|
29
|
Rodrigues CIS, Azevedo RB, Muxfeldt ES. COVID-19 and acute or chronic kidney disease: a crescent learning. J Bras Nefrol 2022; 44:305-307. [PMID: 35920445 PMCID: PMC9518632 DOI: 10.1590/2175-8239-jbn-2022-e005en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 11/24/2022] Open
Affiliation(s)
- Cibele Isaac Saad Rodrigues
- Pontifícia Universidade Católica de São Paulo, Faculdade de Ciências Médicas e da Saúde, Sorocaba, SP, Brazil
| | | | | |
Collapse
|
30
|
Rodrigues CIS, Azevedo RB, Muxfeldt ES. Covid-19 e os rins acometidos com lesão aguda ou crônica: um aprendizado crescente. J Bras Nefrol 2022. [DOI: 10.1590/2175-8239-jbn-2022-e005pt] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
31
|
Abstract
INTRODUCTION : Coronavirus disease 2019 (COVID-19) causes a long-term and persistent condition with clinical features similar to previous virulent outbreaks and other epidemics. Currently, post-COVID syndrome (PCS) is recognized as a new entity in the context of SARS-CoV-2 infection. Though its pathogenesis is not completely understood, persistent inflammation from acute illness and the development of autoimmunity play a critical role in its development. As the pandemic develops, the increasing latent and overt autoimmunity cases indicate that PCS is at the intersection of autoimmunity. AREAS COVERED The mechanisms involved in the emergence of PCS, their similarities with post-viral and post-care syndromes, its inclusion in the spectrum of autoimmunity and possible targets for its treatment. EXPERT OPINION An autoimmune phenomenon plays a major role in most causative theories explaining PCS. Due to the wide scope of symptoms and pathophysiology associated with PCS, there is a need for both PCS definition and classification criteria (including severity scores). Longitudinal and controlled studies are necessary to better understand this new entity, and to confirm that PCS is the chronic phase of COVID-19 as well as to find what additional factors participate into its development. With the high prevalence of COVID-19 cases worldwide, together with the current evidence on latent autoimmunity in PCS, we may observe an increase of autoimmune diseases (ADs) in the coming years. Vaccination's effect on the development of PCS and ADs will also receive attention in the future. Health and social care services need to develop a new framework to deal with PCS.
Collapse
Affiliation(s)
| | - María Herrán
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Santiago Beltrán
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Manuel Rojas
- School of Medicine and Health Sciences, Doctoral Program in Biological and Biomedical Sciences, Universidad del Rosario, Bogota, Colombia.,Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
32
|
Shyfrin SR, Ferren M, Perrin-Cocon L, Espi M, Charmetant X, Brailly M, Decimo D, Iampietro M, Canus L, Horvat B, Lotteau V, Vidalain PO, Thaunat O, Mathieu C. Hamster organotypic kidney culture model of early-stage SARS-CoV-2 infection highlights a two-step renal susceptibility. J Tissue Eng 2022; 13:20417314221122130. [PMID: 36093433 PMCID: PMC9452794 DOI: 10.1177/20417314221122130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/11/2022] [Indexed: 12/16/2022] Open
Abstract
Kidney pathology is frequently reported in patients hospitalized with COVID-19, the pandemic disease caused by the Severe acute respiratory coronavirus 2 (SARS-CoV-2). However, due to a lack of suitable study models, the events occurring in the kidney during the earliest stages of infection remain unknown. We have developed hamster organotypic kidney cultures (OKCs) to study the early stages of direct renal infection. OKCs maintained key renal structures in their native three-dimensional arrangement. SARS-CoV-2 productively replicated in hamster OKCs, initially targeting endothelial cells and later disseminating into proximal tubules. We observed a delayed interferon response, markers of necroptosis and pyroptosis, and an early repression of pro-inflammatory cytokines transcription followed by a strong later upregulation. While it remains an open question whether an active replication of SARS-CoV-2 takes place in the kidneys of COVID-19 patients with AKI, our model provides new insights into the kinetics of SARS-CoV-2 kidney infection and can serve as a powerful tool for studying kidney infection by other pathogens and testing the renal toxicity of drugs.
Collapse
Affiliation(s)
- Sophie R Shyfrin
- CIRI, Centre International de Recherche en Infectiologie, Team Neuro-Invasion, TROpism and VIRal Encephalitis, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France.,CIRI, Centre International de Recherche en Infectiologie, Team Immunobiology of the Viral infections, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Marion Ferren
- CIRI, Centre International de Recherche en Infectiologie, Team Neuro-Invasion, TROpism and VIRal Encephalitis, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France.,CIRI, Centre International de Recherche en Infectiologie, Team Immunobiology of the Viral infections, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Laure Perrin-Cocon
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Maxime Espi
- CIRI, Centre International de Recherche en Infectiologie, Team Normal and pathogenic B cell responses, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Xavier Charmetant
- CIRI, Centre International de Recherche en Infectiologie, Team Normal and pathogenic B cell responses, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Manon Brailly
- CIRI, Centre International de Recherche en Infectiologie, Team Immunobiology of the Viral infections, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Didier Decimo
- CIRI, Centre International de Recherche en Infectiologie, Team Neuro-Invasion, TROpism and VIRal Encephalitis, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France.,CIRI, Centre International de Recherche en Infectiologie, Team Immunobiology of the Viral infections, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Mathieu Iampietro
- CIRI, Centre International de Recherche en Infectiologie, Team Immunobiology of the Viral infections, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Lola Canus
- CIRI, Centre International de Recherche en Infectiologie, Team Neuro-Invasion, TROpism and VIRal Encephalitis, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Branka Horvat
- CIRI, Centre International de Recherche en Infectiologie, Team Immunobiology of the Viral infections, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Vincent Lotteau
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Olivier Thaunat
- CIRI, Centre International de Recherche en Infectiologie, Team Normal and pathogenic B cell responses, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France.,Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France
| | - Cyrille Mathieu
- CIRI, Centre International de Recherche en Infectiologie, Team Neuro-Invasion, TROpism and VIRal Encephalitis, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France.,CIRI, Centre International de Recherche en Infectiologie, Team Immunobiology of the Viral infections, Univ Lyon, Inserm, U1111, CNRS, UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|